EP2984103B1 - Peptides and binding partners therefor - Google Patents

Peptides and binding partners therefor Download PDF

Info

Publication number
EP2984103B1
EP2984103B1 EP14722312.7A EP14722312A EP2984103B1 EP 2984103 B1 EP2984103 B1 EP 2984103B1 EP 14722312 A EP14722312 A EP 14722312A EP 2984103 B1 EP2984103 B1 EP 2984103B1
Authority
EP
European Patent Office
Prior art keywords
peptide
seq
albicans
ece1
infection
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP14722312.7A
Other languages
German (de)
French (fr)
Other versions
EP2984103A1 (en
Inventor
Julian NAGLIK
David MOYES
Shirley TANG
Bernhard HUBE
Duncan WILSON
Sarah HOFS
Jonathan Richardson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Leibniz-Institut fur Naturstoff-Forschung und Infektionsbiologie Ev -Hans-Knoll-Institut- (hki)
Leibniz Institut fuer Naturstoff Forschung und Infektionsbiol eVi
Kings College London
Original Assignee
Leibniz-Institut fur Naturstoff-Forschung und Infektionsbiologie Ev -Hans-Knoll-Institut- (hki)
Leibniz Institut fuer Naturstoff Forschung und Infektionsbiol eVi
Kings College London
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Leibniz-Institut fur Naturstoff-Forschung und Infektionsbiologie Ev -Hans-Knoll-Institut- (hki), Leibniz Institut fuer Naturstoff Forschung und Infektionsbiol eVi, Kings College London filed Critical Leibniz-Institut fur Naturstoff-Forschung und Infektionsbiologie Ev -Hans-Knoll-Institut- (hki)
Publication of EP2984103A1 publication Critical patent/EP2984103A1/en
Application granted granted Critical
Publication of EP2984103B1 publication Critical patent/EP2984103B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/14Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from fungi, algea or lichens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/37Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from fungi
    • C07K14/39Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from fungi from yeasts
    • C07K14/40Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from fungi from yeasts from Candida
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0002Fungal antigens, e.g. Trichophyton, Aspergillus, Candida
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39575Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from other living beings excluding bacteria and viruses, e.g. protozoa, fungi, plants
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6845Methods of identifying protein-protein interactions in protein mixtures
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/20Screening for compounds of potential therapeutic value cell-free systems

Definitions

  • the present invention relates to moieties selected from a specific type of peptide and binding partners such as antibodies therefor, which have applications in diagnosis and therapy. Methods for using these moieties in diagnosis and therapy as well as in processes for screening for therapeutic compounds are also part of the invention.
  • the isolated peptide and specific binding partners therefor are novel and form further aspects of the invention.
  • Candida species are the most common fungal pathogens of humans and the causative agents of oral, gastrointestinal and vaginal candidiasis, giving rise to severe morbidity in millions of individuals worldwide. Vaginal candidiasis affects ⁇ 75% of women at least once during fertile age, equating to ⁇ 30 million infection episodes/year (3x more than tuberculosis and 8x more than HIV: WHO 2007).
  • Candida infections are also the 3 rd most common hospital-acquired bloodstream infection, making Candida species more medically-important than most bacterial infections including Enterococci ( E. coli ) and Pseudomonas spp.
  • Candida pathogens carry an immense health burden and represent a major socio-economic challenge for worldwide communities.
  • Candida albicans is a member of the normal human microbiome. Although typically a commensal of the oral cavity, gastrointestinal and urinogenitary tracts, C. albicans is also an extremely frequent cause of superficial infections such as vaginitis. Moreover, common iatrogenic procedures, such as gastrointestinal surgery, implantation of a central venous catheter or antibiotic treatment are major risk factors for disseminated candidiasis. This form of systemic candidiasis is now the third most common cause of nosocomial bloodstream infections and the mortality of severe sepsis caused by Candida species is over 50% in some patient groups.
  • C. albicans virulence relies on a number of factors, including morphological plasticity, the expression of adhesins and invasins, robust stress responses, immune evasion, metabolic flexibility and nutrient acquisition. However, it remains unclear, how C. albicans causes damage.
  • C. albicans damages host cells have been considered to be multi-factorial, and presumed to rely on a combination of adhesion, invasion, hyphal extension, turgor pressure and the secretion of hydrolytic enzymes.
  • toxin production by C. albicans has long been postulated and the culture supernatants of C. albicans hyphae shown to exhibit haemolytic activity, the mechanism underlying C. albicans ability to lyse host cells has remained elusive. It is clear, however, that hyphae are essential for adhesion, invasion and damage. Thus, damage is caused by hyphae and/or a hyphal associated factor.
  • ECE1 e xtent of c ell e longation 1
  • deletion of ECE1 did not affect hypha formation in C. albicans and phenotypic differences between C. albicans wild type and ECE1 deletion strains have not been reported.
  • ECE1 expression has been used as a marker for hypha formation in multiple independent studies and ECE1 is one of the most strongly expressed genes during hyphal formation.
  • ECE1 is within a small group of core hyphal associated genes. It has previously been reported that recombinant Ecel is processed by the protease, Kex2, at lysine/arginine residues. However, the function of Ecel has never been shown.
  • Ecel plays a key role in host cell activation and damage, as demonstrated by the fact that deletion of ECE1 renders C. albicans unable to damage or activate inflammatory responses in human epithelial cells.
  • a single peptide product of proteolytic processing of Ecel acts as a peptide toxin, in particular, a pore-forming toxin: the first such described in a human fungal pathogen.
  • an optionally labelled peptide comprising SEQ ID NO 1
  • S IIGIIMGIL GNIP QVIQIIMSIV KAFKGNKR SEQ ID NO. 1 or a variant thereof, or an immunogenic fragment of either of these; or a labelled form thereof.
  • the expression 'variant' refers to a peptide sequence in which the amino acid sequence differs from the sequence of SEQ ID NO 1 in that one or more amino acids within the sequence are substituted for other amino acids.
  • the variant produces a biological effect which is similar to that of SEQ ID NO 1.
  • any variant will interact with the surface receptor EGFR (epidermal growth factor receptor) to give rise to activation of an immune response and/or immunoglobulins and in particular antibodies that are produced in response to said variants will cross-react with SEQ ID NO 1.
  • the variant will be damage inducing in host cells, for example by acting as pore forming agents.
  • Amino acid substitutions may be regarded as "conservative" where an amino acid is replaced with a different amino acid in the same class with broadly similar properties.
  • Non-conservative substitutions are where amino acids are replaced with amino acids of a different type or class.
  • Amino acid classes are defined as follows: Class Amino acid examples Nonpolar: A, V, L, I, P, M, F, W Uncharged polar: G, S, T, C, Y, N, Q Acidic: D, E Basic: K, R, H.
  • altering the primary structure of a peptide by a conservative substitution may not significantly alter the activity of that peptide because the side-chain of the amino acid which is inserted into the sequence may be able to form similar bonds and contacts as the side chain of the amino acid which has been substituted out. This is so even when the substitution is in a region which is critical in determining the peptide's conformation.
  • Non-conservative substitutions may also be possible provided that these do not interrupt the function of the peptide and in particular its ability to cross-react with immunoglobulins that react with SEQ ID NO 1.
  • Particular variants may include peptides encoded by orthologues or paralogues of the gene sequence that encodes the peptide of SEQ ID NO 1 in C. albicans.
  • Particular variant sequences are orthologues from C. dubliniensis and C. tropicalis as shown in Figure 2 hereinafter.
  • variants will have amino acid sequences that will be at least 70%, for instance at least 71%, 75%, 79%, 81%, 84%, 87%, 90%, 93% or 96% identical to SEQ ID NO 1.
  • Identity in this context may be determined using the BLASTP computer program with SEQ ID NO 1 as the base sequence.
  • the BLAST software is publicly available at http://blast.ncbi.nlm.nih.gov/Blast.cgi (accessible on 12 March 2009).
  • Variants may also include addition sequences such as tag sequences that may be used for instance in facilitating purification of the peptide or in detection of it.
  • the variant may further comprise an affinity tag such as chitin binding protein (CBP), maltose binding protein (MBP), glutathione-S-transferase (GST), FLAG, myc, biotin or a poly(His) tag as are known in the art.
  • CBP chitin binding protein
  • MBP maltose binding protein
  • GST glutathione-S-transferase
  • FLAG FLAG
  • myc myc
  • biotin biotin
  • poly(His) tag as are known in the art.
  • the variant may comprise a fluorescent protein such as green fluorescent protein (GFP).
  • a chemical label such as a fluorescent or radio label.
  • fluorescent or radio label there are a wide range of such labels available commercially, but examples include fluorescein and derivatives such as fluorescein isothiocyanates (FITC), carboxyfluorescein succinimidyl esters, fluorescein dichlorotriazine (DTAF) and 6-carboxy-4',5'-dichloro-2',7'-dimethoxyfluorescein (JOE), rhodamine and derivatives such as carboxytetramethylrhodamine (TAMRA), tetramethylrhodamine (TMR) and its isothiocyanate derivative (TRITC), sulforhodamine, Texas Red, Rhodamine Red and the dyes available under the trade name Alexa.
  • fluorescein and derivatives such as fluorescein isothiocyanates (FITC), carboxyfluorescein succinimidyl esters, fluorescein dichlorotriazine (
  • fragment refers to any portion of the amino acid sequence of SEQ ID NO 1 which has biological function in common with SEQ ID NO 1. (e.g. damage-inducing, immunomodulatory or is immunogenic, for instance an epitopic fragment) and which reacts with specific binding partners for SEQ ID NO 1.
  • Suitable fragments will include deletion mutants comprising up to 31 amino acids, for example at least 7 amino acids, such as at least 10, for instance at least 15, such as at least 20, more suitably at least 30 amino acids.
  • Seq ID NO 1 includes two segments that contain sequences with high amyloidogenic potential (underlined), which may be involved in cell interactions and activation due to their hydrophobic nature and ability to potentially form ⁇ -helices.
  • Particular variants or fragments of SEQ ID NO 1 will contain at least one of said sequences which are represented below as SEQ ID NO 2 and SEQ ID NO 3 respectively.
  • IIGIIMGIL SEQ ID NO. 2
  • QVIQIIMSIV SEQ ID NO. 3
  • the peptide is a peptide of SEQ ID NO. 1.
  • the peptide is in isolated or purified form and in particular is free of at least some and preferably all other peptides obtainable by Kex2 protease cleavage of the Ecel protein of C. albicans.
  • the peptide may be isolated from C. albicans that has been subject to Kex2 protease digestion, in a particular embodiment, the peptide is prepared synthetically using conventional preparation methods.
  • peptides of the invention and in particular the peptide of SEQ ID NO 1 has been found to be immunogenic.
  • a specific binding partner for a peptide as described above; or a labelled form thereof are immunoglobulins, such as an antibody or a binding fragment thereof.
  • suitable binding fragments of antibodies include Fab, Fab', F(ab)2, F(ab')2 and FV, VH and VK fragments.
  • Antibodies may be polyclonal or monoclonal but in a particular embodiment, the antibodies are monoclonal antibodies.
  • Antibodies can be prepared using conventional methods involving inoculation of an animal such as mouse or guinea pig with a peptide of the first aspect of the invention and harvesting antibodies from the blood thereof. Monoclonal antibodies can be obtained therefrom by fusing antibody producing cells such as spleen cells from the inoculated animal with a hybridoma cell, culturing this cell and harvesting monoclonal antibodies therefrom.
  • the antibody may be chemically labelled to facilitate detection. Suitable labels will be similar to those described above for the peptides of the invention.
  • the antibody may be detectable using a secondary antibody which is modified so as to be detectable using a development reaction, for example because it includes an enzymatic label, as is conventional in the art.
  • a third aspect of the invention provides a peptide as described above for use in therapy, and in particular for the treatment or prevention of infection by Candida albicans.
  • the therapy is for the treatment or prevention of oral, gastrointestinal or mucosal and in particular vaginal infection by Candida albicans.
  • a non-toxic amount of a peptide as described above or a pharmaceutically acceptable composition comprising it is administered to a subject, such as a human or animal.
  • the non-toxic amount is sufficient to produce an immune response that is protective against C. albicans infection.
  • the peptide may be used as a vaccine in a vaccination method.
  • Such a method forms a fourth aspect of the invention.
  • the amount of peptide administered will vary depending upon factors such as the size and health of the patient, the nature of the condition being treated etc. in accordance with normal clinical practice. Typically, a dosage in the range of from 1 ⁇ g-50mg/Kg such as from 1-50 ⁇ g/Kg but in particular from 1-50mg/Kg, for instance from 2-20 mg/Kg, such as from 5-15 mg/Kg would be expected to produce a suitable immune response. Since the peptide of SEQ ID NO 1 has been found to have cytotoxic effects however, care must be taken to ensure that the dosage is sufficient to prime the immune system but not cause unwanted toxic side effects.
  • a variant or fragment of the peptide of SEQ ID NO 1 in the fourth aspect of the invention.
  • full length peptide of SEQ ID NO 1 is capable of inducing cellular lysis and stimulation of the inflammatory response in epithelial cells.
  • These side effects may therefore be avoided by using a variant sequence which is immunogenic, or a fragment of SEQ ID NO 1, in particular one which contains at least SEQ ID NO 2 or SEQ ID NO 3 as described above.
  • the peptide of SEQ ID NO 1 interacts with phospholipids, omission of the dibasic (lysine, arginine) C-terminal head of the peptide may mitigate any unwanted toxic effects.
  • a specific binding partner for a peptide as described above for use in therapy, in particular where the therapy is the treatment or prevention of infection by Candida albicans.
  • the specific binding partner may be used as a vaccine in a vaccination method that may be prophylactic or therapeutic.
  • a vaccination method that may be prophylactic or therapeutic.
  • the amount of specific binding partner administered will vary depending upon factors such as the size and health of the patient, the nature of the condition being treated etc. in accordance with normal clinical practice.
  • a dosage in the range of from 1 to 50 mg/Kg, for instance from 2-30mg/Kg such as from 5-10mg/Kg would produce a suitable therapeutic or protective effect.
  • a seventh aspect of the invention provides a pharmaceutical composition comprising a peptide as described above and a pharmaceutically acceptable carrier.
  • An eighth aspect of the invention provides a pharmaceutical composition comprising a specific binding partner for a peptide as described above and a pharmaceutically acceptable carrier.
  • Suitable pharmaceutical compositions will be in either solid or liquid form. They may be adapted for administration by any convenient route, such as parenteral, oral, vaginal or topical administration or for administration by inhalation or insufflation.
  • the pharmaceutical acceptable carrier may include diluents or excipients which are physiologically tolerable and compatible with the active ingredient.
  • compositions are prepared for injection, for example either subcutaneously or intravenously. They may be liquid solutions or suspensions, or they may be in the form of a solid that is suitable for solution in, or suspension in, liquid prior to injection. Suitable diluents and excipients are, for example, water, saline, dextrose, glycerol, or the like, and combinations thereof. In addition, if desired the compositions may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, stabilizing or pH-buffering agents, and the like.
  • Oral formulations will be in the form of solids or liquids, and may be solutions, syrups, suspensions, tablets, pills, capsules, sustained-release formulations, or powders.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, magnesium carbonate, and the like.
  • Topical formulations will generally take the form of suppositories or intranasal aerosols.
  • traditional binders and excipients may include, for example, polyalkylene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient.
  • Peptides of the first aspect and specific binding partners of the second aspect of the invention may, if required, be produced using recombinant methods.
  • nucleic acids that encode either the peptide or specific binding partner are prepared and these form a ninth aspect of the invention.
  • nucleic acids may be incorporated into a suitable expression vector or plasmid, which is then used to transform a cell and in particular a prokaryotic cell.
  • a suitable nucleic acid that encodes the peptide of SEQ ID NO 1 may correspond to a nucleic acid sequence that occurs naturally in strains of C. albicans strains.
  • the nucleic acid is designed so that it is 'codon optimised' for the expression host being used in the recombinant production method. This should ensure efficient and effective production.
  • the design and preparation of such nucleic acids will be apparent to the skilled person using techniques conventional in the art.
  • a recombinant cell that has been transformed with such a nucleic acid forms a tenth aspect of the invention.
  • Suitable cells will be conventional expression hosts such as E.coli and Pichia pastoris
  • An eleventh aspect of the invention provides a method for producing a peptide as described above, or a specific binding partner therefor, which method comprises culturing a recombinant cell as described above and recovering said peptide or specific binding partner therefrom.
  • the peptides of the first aspect and the specific binding partners of the second aspect of the invention may also be useful in diagnosis of pathogenic invasive C. albicans infection.
  • C. albicans forms part of the commensal flora and so may be found in most individuals at low levels, for example of up to 800 organisms per ml sample.
  • invasive or pathogenic infections may arise, in particular in immunocompromised individuals, and in such cases, the level of C. albicans increases dramatically or there is 'overgrowth'.
  • albicans produces visible symptoms, it would be helpful if tests could be provided that allow for early diagnosis of such infection as this will allow treatment to be started before the infection escalates to a more dangerous systemic infection, or for the diagnosis of invasive pathogenic infection in organs which may not be examined easily such as the intestine.
  • a method for diagnosis of pathogenic invasive infection by C. albicans comprising contacting a sample from an individual with a peptide as described above or a specific binding partner therefor, and detecting whether the sample contains moieties that interact with said peptide or specific binding partner at a sufficient level to indicate a pathogenic invasive infection.
  • Detection of high levels of the peptide of the invention and in particular a peptide of SEQ ID NO 1 in a sample may be indicative of an overgrowth of C. albicans in the individual, leading to the symptoms of candidiasis.
  • This may be determined by using a specific binding partner for the peptide to capture it.
  • the specific binding partner is suitably either directly or indirectly labelled so as to detect or visualise the peptide in a quantitative or semi-quantitative manner.
  • an elevated level of specific binding partner, and in particular, antibodies to the peptide of SEQ ID NO 1 present in the sample can also indicate that an overgrowth of Candida has occurred and that pathogenic invasive infection may result.
  • the diagnostic method will typically utilise a peptide of the invention to bind with antibodies in the sample.
  • the peptide may be immobilised or labelled to assist in the capture and/or detection of the antibodies in the sample.
  • the sample used in the method may be any suitable biological sample including a blood, serum, sputum, saliva or mucosal swabs for instance oral or vaginal swabs, but may also include biopsy samples such as intestinal tissue samples.
  • the levels of peptide or specific binding partner that is indicative of a pathogenic invasive infection or Candida 'overgrowth' rather than the presence of a non-damaging commensal species will vary depending upon factors such as the nature of the sample and the particular patient being examined and the particular technique used to carry out the assay, but will generally be determinable in a particular case in accordance with conventional clinical techniques.
  • levels of C. albicans of up to about 800cells/ml saliva sample may be acceptable as a commensal background, but levels significantly higher than this, in particular at least an order of magnitude greater than this, can be indicative of overgrowth and pathogenic invasive infection. Detection of levels of the peptide of the invention at levels that equate to this cellular population of Candida species would be a cause for concern.
  • Methods include those conventionally known in the art such as immunoassays, for instance, enzyme-linked immuno sorbent assays (ELISA) but may also include multiplex bead assays, immunohistochemistry or immunofluorescence. Such methods are well known in the art.
  • immunoassays for instance, enzyme-linked immuno sorbent assays (ELISA)
  • ELISA enzyme-linked immuno sorbent assays
  • multiplex bead assays immunohistochemistry or immunofluorescence.
  • Kits for carrying out such methods form a further aspect of the invention.
  • the invention further provides a kit for diagnosing a pathogenic invasive infection by C. albicans, said kit comprising a peptide as described above or a specific binding partner therefor, and means for detecting complexes formed between said peptide or specific binding partner and moieties in the sample.
  • detection means will include label means.
  • the label means is suitably one in which the intensity of the signal developed can be related to the concentration of the moiety being detected as this will allow a distinction to be made between levels of C. albicans which is commensal and a level at which disease may result.
  • Suitable label means may be indirect label means which allow signals to develop following a subsequent reaction such as a chemical reaction. These will include enzymatic labels such as peroxidase labels and in particular horseradish peroxidase. Alternatively, labels may include direct labels such as visible plastic labels, fluorescent labels like fluorescein or derivatives thereof, rhodamine or derivatives as described above, as well as radiolabels such as 125 I labels. The labels may be bound to or otherwise associated with the peptide of the invention or the specific binding partner therefor, or they may be linked to a secondary detection moiety such as a secondary antibody or other binding moiety. The precise form the detection means will take will vary depending upon the detection technique employed.
  • detection means may include solid supports such as 96-well plates or beads that may be ferromagnetic in nature.
  • the solid supports will generally have binding moieties for either the peptide of the first aspect or the specific binding partner of the second aspect immobilised thereon.
  • binding moieties are specific for the peptide of the invention, for instance they are specific binding partners of the second aspect of the invention
  • incubation of the sample in contact with the solid support will result in capture of the peptides on the support.
  • a secondary detection antibody that also binds the peptide but carries a label which is either directly or indirectly detectable.
  • Directly detectable labels may comprise visible labels such as fluorescent or radiolabels as described above. Indirect labels may be detected in subsequent reactions such as horseradish peroxidase that is carried by the secondary detection antibody or by a tertiary detection antibody that binds to the immobilised secondary antibody.
  • binding moieties on the solid support specific for a specific binding partner and in particular an antibody to the peptide of the invention, they will generally comprising a peptide of the first aspect of the invention.
  • incubation of the sample in contact with the solid support will result in capture of antibodies to the peptide on the support.
  • the presence of immobilised antibody can similarly be detected by application of a secondary detection antibody that binds the antibody but carries a label which is either directly or indirectly detectable.
  • kits will suitably contain labelled reagents such as suitable stains or dyes that are targeted to the required moiety and in particular the peptide of the invention, as a result of attachment to a suitable specific binding partner.
  • labelled reagents such as suitable stains or dyes that are targeted to the required moiety and in particular the peptide of the invention, as a result of attachment to a suitable specific binding partner.
  • the sample will be prepared in the usual way.
  • a tissue sample is collected, if necessary sectioned and fixed onto a support such as microscope slide whereupon the labelled reagent is administered to the slide so as to visulalise the target moiety and in particular the peptide of SEQ ID NO 1.
  • the kit may comprise one or more additional elements used in immunohistochemical or immunofluorescence method.
  • blocking agents to reduce background signal caused by non-specific binding of the labelled reagent, which blocking agents may include serum, bovine serum albumin or gelatin.
  • the peptide of SEQ ID NO 1 since the peptide of SEQ ID NO 1 has been identified as a cytotoxic element in C. albicans infections, it can be used to develop drugs and other therapies that specifically target this peptide. It may therefore form a useful tool in the screening of antifungal compounds. Screening methods of this type form a further aspect of the invention.
  • Screening methods may take a variety of forms. Typically however, compounds under test will be contacted with a peptide of the invention and in particular a peptide of SEQ ID NO 1 and any interaction between the peptide and the compound will be monitored.
  • the interaction may take place in a vessel or well for instance of a 96-well plate, and the interaction detected using for example, a change in reflectance of polarised light caused by binding.
  • the interaction may take place at a surface at which one of the peptide or the target reagent is immobilised and under conditions where interaction may be detected using techniques such as surface plasmon resonance and the like.
  • C. albicans gene deletion mutants lacking ECE1, as well as revertant mutant strains were created.
  • C. albicans gene deletion cassettes were generated using the PCR-based method described by Dalle F et al. Cellular Microbiology 2010; 12:248-71 .
  • Primers ECE1-FG and ECE1-RG as shown below as SEQ ID NO 4 and 5 respectively, were used to amplify the HIS1 and ARG4 markers from plasmids pFA-HIS1 and pFA-ARG4, respectively.
  • the C. albicans strain BWP17 Wachtler B, et al.
  • ECE1 open reading frame plus upstream and downstream intergenic regions were amplified with primers ECE1-RecF3k (SEQ ID NO 6) and ECE1-RecR (SEQ ID NO 7) and cloned into plasmid CIp10 at Mlu I and Sal I sites, yielding plasmid CIp10-ECE1.
  • This plasmid was transfomed into the uridine auxotrophic ece1 ⁇ - deletion strain, yielding the ece1 ⁇ / ⁇ - ECE1 complemented strain.
  • LDH lactate dehydrogenase
  • Ecel can be proteolytically processed by Kex2 into eight peptide fragments ( O. Bader et al. BMC Microbiology 2008, 8:115 ).
  • Figure 2 illustrates the structure of Ecel and the proposed resultant peptide fragments following proteolytic processing as well as its phylogenic relationship with orthologues in C. dubliniensis and C. tropicalis (note that ECE1 orthologues are only found in these two fungal species).
  • Each of the individual C. albicans peptides were synthesised by ProteoGenix using fmoc solid phase peptide synthesis technology.
  • Example 1 The method of Example 1 was then repeated using a 1:1 mixture of peptides (1-7) of Figure 2 on human TR146 oral epithelial cells at various concentrations (250 ⁇ g/ml, 125 ⁇ g/ml, 62.5 ⁇ g/ml, 31.25 ⁇ g/ml, 15.625 ⁇ g/ml and 7.8 ⁇ g/ml of each individual peptide within the mixture) or peptide 3 (SEQ ID NO 1) alone in the presence of the ece1 ⁇ mutant C. albicans. Peptide 8 could not be included, as this peptide was insoluble. After incubations for 24 hours, the damage was assessed by measuring LDH release as before. The results are shown in Figure 3A . This shows that peptide 3 (designated Ece1-III in the Figure) produced similar levels of damage to the combination at a concentration of 250 ⁇ g/ml.
  • Each individual peptide (at a final concentration of 30 ⁇ g/ml) was incubated with 1 ⁇ 10 7 human erythrocytes in a volume of 150 ⁇ l. After incubation for 1 hour at 37°C, haemolysis was assayed by measuring haemoglobin release, as indicated by absorbance at 541nm, normalised to a 100% lysis-water control.
  • Pore-forming toxins are generally strong activators of inflammatory responses.
  • recognition of C. albicans hyphae resulted in the activation of the mitogen-activated protein kinase (MAPK) p38 signalling pathway and the c-Fos transcription factor, which in turn activates proinflammatory cytokine production ( Moyes DL, et al.: Cell Host Microbe 2010, 8:225-235 ,).
  • An extensive screen of >100 C . albicans mutants was carried out using protocols described in this reference and also Moyes DL, et al. PLoS ONE 2011, 6:e26580 ; Murciano C. et al.
  • Ece1-III As the region of Ecel responsible for cellular lysis and the activation of immune responses, the applicants next investigated the precise functional requirements of the Ece1-III amino acid sequence ( Figure 8A ). Accordingly, individual peptide fragments corresponding to internal regions of Ece1-III were constructed (WTSA peptides 1-6 (SEQ ID NOS 8-11, 3 and 12 respectively: Figure 8B ). The sequence of each WTSA peptide was chosen to facilitate a detailed examination of each of the internal amyloidogenic regions of Ece1-III, either alone (WTSA peptides 1, 2, 4 and 5), or in combination (WTSA 3).
  • the p38/c-Fos pathway is activated via the interaction of C. albicans hyphae with the surface receptor EGFR (epidermal growth factor receptor), as blocking EGFR activation also blocks c-Fos activation ( Figure 11A ).
  • Biacore binding assays show that Ece1-III (of SEQ ID NO 1), but not other parts of the Ecel protein, directly interact with EGFR with high affinity ( Figure 11B ) and that blocking EGFR abolishes the ability of Ece1-III to activate c-Fos ( Figure 11C ). Furthermore, blocking EGFR also reduced/abolished activation of all three MAPK signalling pathways (p38, JNK and ERK1/2) ( Fig.
  • Ece1-III consists of a predicted alpha helix with a dibasic (lysine, arginine) C-terminal head, the applicants predicted that these cationic, positively charged residues may interact with negatively charged phospholipids present in target membranes.
  • Ece1-III was used to probe a phospholipid array (PIP Strip membrane). Binding was assessed by hybridisation with anti-Ece1-III antibody, followed by detection with anti-rabbit antibody conjugated to horse radish peroxidase. The results are shown in Figure 13 .
  • Ece1-III interacts with Phosphatidylinositol-3-phosphate, Phosphatidylinositol-4-phosphate, Phosphatidylinositol-5-phosphate, Phosphatidylinositol-3,4-phosphate, Phosphatidylinositol-3,5 -phosphate, Phosphatidylinositol-4,5-phosphate, Phosphatidylinositol-3,4,5-phosphate, Phosphatidic acid and Phosphatidylserine, with binding to Phosphatidylinositol-3-phosphate being particularly robust ( Figure 13 ).
  • Ece1-III did not interact with Lysophosphatidic acid, Lysophosphatidylcholine, Phosphatidylinositol, Phosphatidylethanolamine, Phosphatidylcholine or Sphingosine-1-phosphate. Therefore, insertion of Ece1-III into target membranes (pore formation) may be mediated by interactions with phospholipids.
  • Ece1-III Given the importance of Ece1-III in killing epithelial cells, the applicants examined whether it also plays a role in immune escape of C. albicans from macrophages.
  • C. albicans yeast cells are phagocytosed by macrophages, but rapidly germinate and escape the macrophage via hypha formation.
  • Ecel is involved in this process, wild type, ece1 ⁇ , and ece1 ⁇ / ⁇ - ECE1 cells were coincubated with macrophages and the following parameters measured: phagocytosis rates, length of hyphae, escape from macrophages, and damage of macrophages.
  • mice were first immunosuppressed with cortisone acetate prior to administration of wild-type (parental) C. albicans (BWP17), a ece1 ⁇ null mutant and ece1 ⁇ / ⁇ - ECE1 revertant strain into the oral cavity. After four days, fungal burdens, fungal invasion, tissue damage and immune cell recruitment was determined in tongue tissues.
  • mice infected with the ece1 ⁇ null mutant were found in mice infected with the ece1 ⁇ null mutant as compared with the wild-type (BWP17) or the ece1 ⁇ / ⁇ - ECE1 revertant strains, and in many cases the ece1 ⁇ null mutant could not be recovered from mice ( Figure 16 A) .
  • Histological analysis of mouse tongues in both the wild-type (BWP17) and ece1 ⁇ / ⁇ - ECE1 revertant strain showed multiple foci of infection that were associated with inflammatory immune cells (neutrophils) and extensive local tissue damage ( Figure 16 B-D ).
  • mice infected with the ece1 ⁇ null mutant showed no evidence of any foci of invasion, tissue damage or inflammation and no evidence of C. albicans was detectable ( Figure 16 E) . Therefore, Ecel is essential for successful mucosal infection, damage induction and immune activation in this model.
  • the applicants also determined the effect of administering full length His-tagged Ecelp and Ece1-III in a Drosophila melanogaster (fruit fly) model. All flies injected with full length His-tagged Ecelp and Ece1-III displayed signs of paralysis, with only 17.5% and 10% recovering after 24 hours, as compared with the negative control Ecel-II (peptide 2) which showed no signs of paralysis ( Figure 17 ). This indicates that Ece1-III may have a direct or indirect neurotoxic effect.
  • Ecel and its active component Ece1-III
  • Ece1-III has a dual functional role, by acting both as a pore-forming toxin that damages host cells and as an activator of immune responses. Damage of host cells and uncontrolled immune activation are the hallmarks of several diseases caused by C. albicans, which can often lead to death (45- 76% mortality) during systemic infections. Therefore, neutralisation of Ece1-III will prevent not only host damage during C. albicans infections but also deleterious inflammatory responses. This indicates that Ece1-III represents a new therapeutic target to treat C. albicans infections.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Mycology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Analytical Chemistry (AREA)
  • Botany (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Description

  • The present invention relates to moieties selected from a specific type of peptide and binding partners such as antibodies therefor, which have applications in diagnosis and therapy. Methods for using these moieties in diagnosis and therapy as well as in processes for screening for therapeutic compounds are also part of the invention. The isolated peptide and specific binding partners therefor are novel and form further aspects of the invention.
  • Background to the Invention
  • Candida species are the most common fungal pathogens of humans and the causative agents of oral, gastrointestinal and vaginal candidiasis, giving rise to severe morbidity in millions of individuals worldwide. Vaginal candidiasis affects ∼75% of women at least once during fertile age, equating to ∼30 million infection episodes/year (3x more than tuberculosis and 8x more than HIV: WHO 2007). Candida infections are also the 3rd most common hospital-acquired bloodstream infection, making Candida species more medically-important than most bacterial infections including Enterococci (E. coli) and Pseudomonas spp. Systemic candidiasis is fatal (30-50% mortality) with ∼300,000 cases/year of candidaemia, equating to ∼100,000 deaths/year. Furthermore, Candida infections are the most common oral manifestation of HIV infection, with 50% of HIV+ patients and 90% AIDS patients suffering from oral candidiasis. With ∼4 million cases of HIV/year, this equates to ∼2 million oral candidiasis cases/year. Indeed, one of the biggest killers of the immunocompromised population is fungal infection. In the USA, yearly healthcare costs for fungal infections are $2.6 billion, of which Candida infections account for $1.8 billion. EU healthcare costs are estimated to be similar. Therefore, Candida pathogens carry an immense health burden and represent a major socio-economic challenge for worldwide communities.
  • Candida albicans is a member of the normal human microbiome. Although typically a commensal of the oral cavity, gastrointestinal and urinogenitary tracts, C. albicans is also an extremely frequent cause of superficial infections such as vaginitis. Moreover, common iatrogenic procedures, such as gastrointestinal surgery, implantation of a central venous catheter or antibiotic treatment are major risk factors for disseminated candidiasis. This form of systemic candidiasis is now the third most common cause of nosocomial bloodstream infections and the mortality of severe sepsis caused by Candida species is over 50% in some patient groups.
  • Identifying new epithelial and fungal targets that stimulate protective host immunity will ultimately have major implications for global health.
  • C. albicans virulence relies on a number of factors, including morphological plasticity, the expression of adhesins and invasins, robust stress responses, immune evasion, metabolic flexibility and nutrient acquisition. However, it remains unclear, how C. albicans causes damage.
  • The mechanisms by which C. albicans damages host cells have been considered to be multi-factorial, and presumed to rely on a combination of adhesion, invasion, hyphal extension, turgor pressure and the secretion of hydrolytic enzymes. Although toxin production by C. albicans has long been postulated and the culture supernatants of C. albicans hyphae shown to exhibit haemolytic activity, the mechanism underlying C. albicans ability to lyse host cells has remained elusive. It is clear, however, that hyphae are essential for adhesion, invasion and damage. Thus, damage is caused by hyphae and/or a hyphal associated factor.
  • The gene ECE1 (extent of cell elongation 1) was first identified two decades ago due to its heightened expression during hypha formation. However, deletion of ECE1 did not affect hypha formation in C. albicans and phenotypic differences between C. albicans wild type and ECE1 deletion strains have not been reported. Despite this, ECE1 expression has been used as a marker for hypha formation in multiple independent studies and ECE1 is one of the most strongly expressed genes during hyphal formation. In fact, ECE1 is within a small group of core hyphal associated genes. It has previously been reported that recombinant Ecel is processed by the protease, Kex2, at lysine/arginine residues. However, the function of Ecel has never been shown.
  • Currently, the gold standard for treating fungal infections is the use of antifungal drugs, which predominantly target either the cell wall (echinocandins), cell membrane (polyenes) or ergosterol synthesis (azoles). However, the continuing increase in Candida infections together with increasing drug resistance highlights the need to discover new and better agents that target either (i) epithelial processes that recognise and normally restrict these potentially life-threatening pathogens to mucosal surfaces or (ii) fungal determinants that promote infection and/or mediate immune activation and protective immunity. Despite this, there are no vaccine candidates or immune-based intervention strategies for combating Candida infections. Likewise, there are no commercial drugs that specifically target mucosal fungal infections.
  • Summary of the Invention
  • The applicants have found that Ecel plays a key role in host cell activation and damage, as demonstrated by the fact that deletion of ECE1 renders C. albicans unable to damage or activate inflammatory responses in human epithelial cells. Moreover, the applicants have demonstrated that a single peptide product of proteolytic processing of Ecel acts as a peptide toxin, in particular, a pore-forming toxin: the first such described in a human fungal pathogen. By targeting this single peptide it is possible to prevent both host damage (fungus driven) and manipulate immunity to induce protection (host driven). To date, no other Candida protein exhibits these phenotypes and no other approach can address both fungal and host aspects.
  • This gives rise to a number of diagnostic and therapeutic applications for this peptide and also for specific binding partners for it, as well as for the discovery and development of anti-fungal compounds that target this specific peptide.
  • According to a first aspect of the present invention there is provided an optionally labelled peptide comprising SEQ ID NO 1
  • SIIGIIMGILGNIPQVIQIIMSIVKAFKGNKR (SEQ ID NO. 1) or a variant thereof, or an immunogenic fragment of either of these; or a labelled form thereof.
  • As used herein, the expression 'variant' refers to a peptide sequence in which the amino acid sequence differs from the sequence of SEQ ID NO 1 in that one or more amino acids within the sequence are substituted for other amino acids. However, the variant produces a biological effect which is similar to that of SEQ ID NO 1. In particular, any variant will interact with the surface receptor EGFR (epidermal growth factor receptor) to give rise to activation of an immune response and/or immunoglobulins and in particular antibodies that are produced in response to said variants will cross-react with SEQ ID NO 1. Alternatively or additionally, the variant will be damage inducing in host cells, for example by acting as pore forming agents.
  • Amino acid substitutions may be regarded as "conservative" where an amino acid is replaced with a different amino acid in the same class with broadly similar properties. Non-conservative substitutions are where amino acids are replaced with amino acids of a different type or class.
  • Amino acid classes are defined as follows:
    Class Amino acid examples
    Nonpolar: A, V, L, I, P, M, F, W
    Uncharged polar: G, S, T, C, Y, N, Q
    Acidic: D, E
    Basic: K, R, H.
  • As is well known to those skilled in the art, altering the primary structure of a peptide by a conservative substitution may not significantly alter the activity of that peptide because the side-chain of the amino acid which is inserted into the sequence may be able to form similar bonds and contacts as the side chain of the amino acid which has been substituted out. This is so even when the substitution is in a region which is critical in determining the peptide's conformation.
  • Non-conservative substitutions may also be possible provided that these do not interrupt the function of the peptide and in particular its ability to cross-react with immunoglobulins that react with SEQ ID NO 1.
  • Broadly speaking, fewer non-conservative substitutions will be possible without altering the biological activity of the polypeptides.
  • Particular variants may include peptides encoded by orthologues or paralogues of the gene sequence that encodes the peptide of SEQ ID NO 1 in C. albicans. Particular variant sequences are orthologues from C. dubliniensis and C. tropicalis as shown in Figure 2 hereinafter.
  • In general, variants will have amino acid sequences that will be at least 70%, for instance at least 71%, 75%, 79%, 81%, 84%, 87%, 90%, 93% or 96% identical to SEQ ID NO 1. Identity in this context may be determined using the BLASTP computer program with SEQ ID NO 1 as the base sequence. The BLAST software is publicly available at http://blast.ncbi.nlm.nih.gov/Blast.cgi (accessible on 12 March 2009).
  • Variants may also include addition sequences such as tag sequences that may be used for instance in facilitating purification of the peptide or in detection of it. Thus for instance, the variant may further comprise an affinity tag such as chitin binding protein (CBP), maltose binding protein (MBP), glutathione-S-transferase (GST), FLAG, myc, biotin or a poly(His) tag as are known in the art. In another embodiment, the variant may comprise a fluorescent protein such as green fluorescent protein (GFP).
  • When the peptide is for use in diagnostics or screening of anti-fungal proteins, it may be helpful to label the peptide, for example with a chemical label such as a fluorescent or radio label. There are a wide range of such labels available commercially, but examples include fluorescein and derivatives such as fluorescein isothiocyanates (FITC), carboxyfluorescein succinimidyl esters, fluorescein dichlorotriazine (DTAF) and 6-carboxy-4',5'-dichloro-2',7'-dimethoxyfluorescein (JOE), rhodamine and derivatives such as carboxytetramethylrhodamine (TAMRA), tetramethylrhodamine (TMR) and its isothiocyanate derivative (TRITC), sulforhodamine, Texas Red, Rhodamine Red and the dyes available under the trade name Alexa.
  • These labels may be incorporated into the peptide using conventional methods.
  • The term "fragment" as used herein refers to any portion of the amino acid sequence of SEQ ID NO 1 which has biological function in common with SEQ ID NO 1. (e.g. damage-inducing, immunomodulatory or is immunogenic, for instance an epitopic fragment) and which reacts with specific binding partners for SEQ ID NO 1. Suitable fragments will include deletion mutants comprising up to 31 amino acids, for example at least 7 amino acids, such as at least 10, for instance at least 15, such as at least 20, more suitably at least 30 amino acids.
  • Notably, Seq ID NO 1 includes two segments that contain sequences with high amyloidogenic potential (underlined), which may be involved in cell interactions and activation due to their hydrophobic nature and ability to potentially form α-helices. Particular variants or fragments of SEQ ID NO 1 will contain at least one of said sequences which are represented below as SEQ ID NO 2 and SEQ ID NO 3 respectively.
    IIGIIMGIL (SEQ ID NO. 2)
    QVIQIIMSIV (SEQ ID NO. 3)
  • In a particular embodiment however, the peptide is a peptide of SEQ ID NO. 1. The peptide is in isolated or purified form and in particular is free of at least some and preferably all other peptides obtainable by Kex2 protease cleavage of the Ecel protein of C. albicans. Although the peptide may be isolated from C. albicans that has been subject to Kex2 protease digestion, in a particular embodiment, the peptide is prepared synthetically using conventional preparation methods.
  • As discussed above, peptides of the invention and in particular the peptide of SEQ ID NO 1 has been found to be immunogenic. Thus in a second aspect of the present invention there is provided a specific binding partner for a peptide as described above; or a labelled form thereof. Particular specific binding partners are immunoglobulins, such as an antibody or a binding fragment thereof. Examples of suitable binding fragments of antibodies include Fab, Fab', F(ab)2, F(ab')2 and FV, VH and VK fragments.
  • Antibodies may be polyclonal or monoclonal but in a particular embodiment, the antibodies are monoclonal antibodies.
  • Antibodies can be prepared using conventional methods involving inoculation of an animal such as mouse or guinea pig with a peptide of the first aspect of the invention and harvesting antibodies from the blood thereof. Monoclonal antibodies can be obtained therefrom by fusing antibody producing cells such as spleen cells from the inoculated animal with a hybridoma cell, culturing this cell and harvesting monoclonal antibodies therefrom.
  • Where the antibody is used in diagnostic or screening applications, it may be chemically labelled to facilitate detection. Suitable labels will be similar to those described above for the peptides of the invention. However, the antibody may be detectable using a secondary antibody which is modified so as to be detectable using a development reaction, for example because it includes an enzymatic label, as is conventional in the art.
  • The peptide of the invention has therapeutic applications. Therefore, a third aspect of the invention provides a peptide as described above for use in therapy, and in particular for the treatment or prevention of infection by Candida albicans. In particular, the therapy is for the treatment or prevention of oral, gastrointestinal or mucosal and in particular vaginal infection by Candida albicans.
  • In order to achieve these therapeutic effects, a non-toxic amount of a peptide as described above or a pharmaceutically acceptable composition comprising it is administered to a subject, such as a human or animal. The non-toxic amount is sufficient to produce an immune response that is protective against C. albicans infection. Thus the peptide may be used as a vaccine in a vaccination method. Such a method forms a fourth aspect of the invention.
  • The amount of peptide administered will vary depending upon factors such as the size and health of the patient, the nature of the condition being treated etc. in accordance with normal clinical practice. Typically, a dosage in the range of from 1µg-50mg/Kg such as from 1-50µg/Kg but in particular from 1-50mg/Kg, for instance from 2-20 mg/Kg, such as from 5-15 mg/Kg would be expected to produce a suitable immune response. Since the peptide of SEQ ID NO 1 has been found to have cytotoxic effects however, care must be taken to ensure that the dosage is sufficient to prime the immune system but not cause unwanted toxic side effects.
  • One way of avoiding such side effects is to use a variant or fragment of the peptide of SEQ ID NO 1 in the fourth aspect of the invention. As described hereinbelow, the applicants have found that full length peptide of SEQ ID NO 1 is capable of inducing cellular lysis and stimulation of the inflammatory response in epithelial cells. These side effects may therefore be avoided by using a variant sequence which is immunogenic, or a fragment of SEQ ID NO 1, in particular one which contains at least SEQ ID NO 2 or SEQ ID NO 3 as described above. Furthermore, the applicants have found that the peptide of SEQ ID NO 1 interacts with phospholipids, omission of the dibasic (lysine, arginine) C-terminal head of the peptide may mitigate any unwanted toxic effects.
  • An alternative way of avoiding unwanted side effects is to use the specific binding partners and in particular the antibodies of the second aspect of the invention in a passive immunisation regime. Thus in a fifth aspect of the invention, there is provided a specific binding partner for a peptide as described above for use in therapy, in particular where the therapy is the treatment or prevention of infection by Candida albicans.
  • As before, in order to achieve these effects, an effective amount of the specific binding partner and in particular the antibody, or a pharmaceutical composition containing it is administered to a subject in need thereof. Thus the specific binding partner may be used as a vaccine in a vaccination method that may be prophylactic or therapeutic. Such a method forms a sixth aspect of the invention.
  • Again, the amount of specific binding partner administered will vary depending upon factors such as the size and health of the patient, the nature of the condition being treated etc. in accordance with normal clinical practice. Typically, a dosage in the range of from 1 to 50 mg/Kg, for instance from 2-30mg/Kg such as from 5-10mg/Kg would produce a suitable therapeutic or protective effect.
  • For administration to subjects, the peptide or its specific binding partner is suitably administered in the form of a pharmaceutical composition. Thus a seventh aspect of the invention provides a pharmaceutical composition comprising a peptide as described above and a pharmaceutically acceptable carrier.
  • An eighth aspect of the invention provides a pharmaceutical composition comprising a specific binding partner for a peptide as described above and a pharmaceutically acceptable carrier.
  • Suitable pharmaceutical compositions will be in either solid or liquid form. They may be adapted for administration by any convenient route, such as parenteral, oral, vaginal or topical administration or for administration by inhalation or insufflation. The pharmaceutical acceptable carrier may include diluents or excipients which are physiologically tolerable and compatible with the active ingredient.
  • Parenteral compositions are prepared for injection, for example either subcutaneously or intravenously. They may be liquid solutions or suspensions, or they may be in the form of a solid that is suitable for solution in, or suspension in, liquid prior to injection. Suitable diluents and excipients are, for example, water, saline, dextrose, glycerol, or the like, and combinations thereof. In addition, if desired the compositions may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, stabilizing or pH-buffering agents, and the like.
  • Oral formulations will be in the form of solids or liquids, and may be solutions, syrups, suspensions, tablets, pills, capsules, sustained-release formulations, or powders. Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, magnesium carbonate, and the like.
  • Topical formulations will generally take the form of suppositories or intranasal aerosols. For suppositories, traditional binders and excipients may include, for example, polyalkylene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient.
  • Peptides of the first aspect and specific binding partners of the second aspect of the invention, may, if required, be produced using recombinant methods. For this purpose, nucleic acids that encode either the peptide or specific binding partner are prepared and these form a ninth aspect of the invention.
  • Such nucleic acids may be incorporated into a suitable expression vector or plasmid, which is then used to transform a cell and in particular a prokaryotic cell. A suitable nucleic acid that encodes the peptide of SEQ ID NO 1 may correspond to a nucleic acid sequence that occurs naturally in strains of C. albicans strains. However, in a particular embodiment, the nucleic acid is designed so that it is 'codon optimised' for the expression host being used in the recombinant production method. This should ensure efficient and effective production. The design and preparation of such nucleic acids will be apparent to the skilled person using techniques conventional in the art.
  • A recombinant cell that has been transformed with such a nucleic acid forms a tenth aspect of the invention.
  • Suitable cells will be conventional expression hosts such as E.coli and Pichia pastoris An eleventh aspect of the invention provides a method for producing a peptide as described above, or a specific binding partner therefor, which method comprises culturing a recombinant cell as described above and recovering said peptide or specific binding partner therefrom.
  • However, the peptides of the first aspect and the specific binding partners of the second aspect of the invention may also be useful in diagnosis of pathogenic invasive C. albicans infection. C. albicans forms part of the commensal flora and so may be found in most individuals at low levels, for example of up to 800 organisms per ml sample. However, as discussed previously, invasive or pathogenic infections may arise, in particular in immunocompromised individuals, and in such cases, the level of C. albicans increases dramatically or there is 'overgrowth'. Although in many cases, pathogenic invasive oral or vaginal infection of C. albicans produces visible symptoms, it would be helpful if tests could be provided that allow for early diagnosis of such infection as this will allow treatment to be started before the infection escalates to a more dangerous systemic infection, or for the diagnosis of invasive pathogenic infection in organs which may not be examined easily such as the intestine.
  • In an eleventh aspect of the invention, there is provided a method for diagnosis of pathogenic invasive infection by C. albicans, said method comprising contacting a sample from an individual with a peptide as described above or a specific binding partner therefor, and detecting whether the sample contains moieties that interact with said peptide or specific binding partner at a sufficient level to indicate a pathogenic invasive infection.
  • Detection of high levels of the peptide of the invention and in particular a peptide of SEQ ID NO 1 in a sample may be indicative of an overgrowth of C. albicans in the individual, leading to the symptoms of candidiasis. This may be determined by using a specific binding partner for the peptide to capture it. The specific binding partner is suitably either directly or indirectly labelled so as to detect or visualise the peptide in a quantitative or semi-quantitative manner.
  • Similarly, an elevated level of specific binding partner, and in particular, antibodies to the peptide of SEQ ID NO 1 present in the sample can also indicate that an overgrowth of Candida has occurred and that pathogenic invasive infection may result. In this case, the diagnostic method will typically utilise a peptide of the invention to bind with antibodies in the sample. The peptide may be immobilised or labelled to assist in the capture and/or detection of the antibodies in the sample.
  • The sample used in the method may be any suitable biological sample including a blood, serum, sputum, saliva or mucosal swabs for instance oral or vaginal swabs, but may also include biopsy samples such as intestinal tissue samples.
  • The levels of peptide or specific binding partner that is indicative of a pathogenic invasive infection or Candida 'overgrowth' rather than the presence of a non-damaging commensal species will vary depending upon factors such as the nature of the sample and the particular patient being examined and the particular technique used to carry out the assay, but will generally be determinable in a particular case in accordance with conventional clinical techniques. Typically however, levels of C. albicans of up to about 800cells/ml saliva sample may be acceptable as a commensal background, but levels significantly higher than this, in particular at least an order of magnitude greater than this, can be indicative of overgrowth and pathogenic invasive infection. Detection of levels of the peptide of the invention at levels that equate to this cellular population of Candida species would be a cause for concern.
  • Methods include those conventionally known in the art such as immunoassays, for instance, enzyme-linked immuno sorbent assays (ELISA) but may also include multiplex bead assays, immunohistochemistry or immunofluorescence. Such methods are well known in the art.
  • Kits for carrying out such methods form a further aspect of the invention. Thus the invention further provides a kit for diagnosing a pathogenic invasive infection by C. albicans, said kit comprising a peptide as described above or a specific binding partner therefor, and means for detecting complexes formed between said peptide or specific binding partner and moieties in the sample. The nature of the detection means will vary depending upon the particular technique being employed. In general, detection means will include label means. The label means is suitably one in which the intensity of the signal developed can be related to the concentration of the moiety being detected as this will allow a distinction to be made between levels of C. albicans which is commensal and a level at which disease may result.
  • Suitable label means may be indirect label means which allow signals to develop following a subsequent reaction such as a chemical reaction. These will include enzymatic labels such as peroxidase labels and in particular horseradish peroxidase. Alternatively, labels may include direct labels such as visible plastic labels, fluorescent labels like fluorescein or derivatives thereof, rhodamine or derivatives as described above, as well as radiolabels such as 125I labels. The labels may be bound to or otherwise associated with the peptide of the invention or the specific binding partner therefor, or they may be linked to a secondary detection moiety such as a secondary antibody or other binding moiety. The precise form the detection means will take will vary depending upon the detection technique employed.
  • For instance for immunoassays such as ELISA assays, detection means may include solid supports such as 96-well plates or beads that may be ferromagnetic in nature. In such cases, the solid supports will generally have binding moieties for either the peptide of the first aspect or the specific binding partner of the second aspect immobilised thereon.
  • Where the binding moieties are specific for the peptide of the invention, for instance they are specific binding partners of the second aspect of the invention, incubation of the sample in contact with the solid support will result in capture of the peptides on the support. Once the sample has then been removed from the support for example by washing, the presence of immobilised peptide can be detected by application of a secondary detection antibody that also binds the peptide but carries a label which is either directly or indirectly detectable. Directly detectable labels may comprise visible labels such as fluorescent or radiolabels as described above. Indirect labels may be detected in subsequent reactions such as horseradish peroxidase that is carried by the secondary detection antibody or by a tertiary detection antibody that binds to the immobilised secondary antibody.
  • Similarly where the binding moieties on the solid support specific for a specific binding partner and in particular an antibody to the peptide of the invention, they will generally comprising a peptide of the first aspect of the invention. In this case, incubation of the sample in contact with the solid support will result in capture of antibodies to the peptide on the support. Once the sample has then been removed from the support for example by washing, the presence of immobilised antibody can similarly be detected by application of a secondary detection antibody that binds the antibody but carries a label which is either directly or indirectly detectable.
  • For immunohistochemical or immunofluorescence detection methods, kits will suitably contain labelled reagents such as suitable stains or dyes that are targeted to the required moiety and in particular the peptide of the invention, as a result of attachment to a suitable specific binding partner. In use, the sample will be prepared in the usual way. In particular, a tissue sample is collected, if necessary sectioned and fixed onto a support such as microscope slide whereupon the labelled reagent is administered to the slide so as to visulalise the target moiety and in particular the peptide of SEQ ID NO 1. The kit may comprise one or more additional elements used in immunohistochemical or immunofluorescence method. These include the slide itself, fixing agent such as paraformaldehyde, detergent such as Triton® detergent for reducing surface tension and facilitating coverage by the labelled reagent, and blocking agents to reduce background signal caused by non-specific binding of the labelled reagent, which blocking agents may include serum, bovine serum albumin or gelatin.
  • In addition, since the peptide of SEQ ID NO 1 has been identified as a cytotoxic element in C. albicans infections, it can be used to develop drugs and other therapies that specifically target this peptide. It may therefore form a useful tool in the screening of antifungal compounds. Screening methods of this type form a further aspect of the invention.
  • Screening methods may take a variety of forms. Typically however, compounds under test will be contacted with a peptide of the invention and in particular a peptide of SEQ ID NO 1 and any interaction between the peptide and the compound will be monitored. The interaction may take place in a vessel or well for instance of a 96-well plate, and the interaction detected using for example, a change in reflectance of polarised light caused by binding. Alternatively, the interaction may take place at a surface at which one of the peptide or the target reagent is immobilised and under conditions where interaction may be detected using techniques such as surface plasmon resonance and the like.
  • Detailed Description of the Invention
  • The invention will now be particularly described by way of example with reference to the accompanying diagrams which are summarised as follows. However, it will be apparent to one skilled in the art that the specific details are not required in order to practice the invention. The following descriptions of specific embodiments of the present invention are presented for purposes of illustration and description. They are not intended to be exhaustive of or to limit the invention to the precise forms disclosed. Many modifications and variations are possible in view of the above teachings. The embodiments are shown and described in order to best explain the principles of the invention and its practical applications, to thereby enable others skilled in the art to best utilize the invention and various embodiments with various modifications as are suited to the particular use contemplated.
    • Figure 1 is a series of graph showing the results of an experiment to determine whether ECE1 is required for damage of a variety of host cells. Oral TR146 (A), gastrointestinal Caco (B) and vaginal (C) epithelial cells were infected with C. albicans wild type (wt), ece1Δ or ece1Δ,ECE1 strains for 24 hours and damage assayed by LDH release as described below.
    • Figure 2 shows the structure of Ecel and phylogeny with other species. It shows the alignment of Ecel from C. albicans (SEQ ID NO 13) and orthologues in C. dubliniensis (SEQ ID NO 14) and C. tropicalis (SEQ ID NO 15) (which are the only three sequenced fungal species that encode Ecel). The bars below the alignment indicate that the peptides resulted from Kex2 digestion of the CaEce1. Only peptide 3 of C. albicans, C. dubliniensis and C. tropicalis form transmembrane α-helical structures.
    • Figure 3 is a series of graphs showing the results of an experiment to demonstrate that a peptide of the invention is sufficient to lyse host epithelial cells. Mixtures of Ecel peptides at different concentrations or the peptide of SEQ ID NO 1 alone were added to TR146 oral epithelial cells in the presence of the ece1Δ mutant, incubated for 24 hours and damage assessed by measuring LDH release (A). Subsequently the difference Ecel peptides were added alone (without C. ablicans cells) to TR146 (B) and vaginal A431(C) epithelia, incubated for 24 hours and damage assessed by measuring LDH release as described below.
    • Figure 4 is a graph showing the results of an experiment to determine which of the Ecel peptides are haemolytic toxins where the peptide of SEQ ID NO 1 is peptide 3. Individual peptides were incubated with human erythrocytes for 1 hour and heamolysis assayed by measuring haemoglobin release (as measured by absorbance at 541nm, normalised to a 100% lysis control).
    • Figure 5 shows the results obtained by scanning electron microscopy of an experiment to determine whether the peptide of SEQ ID NO 1 forms pores in epithelial membranes. Oral epithelial (TR146) monolayers were incubated with 5µg/ml peptide 3 (SEQ ID NO 1) for 5 hours and pore formation assessed by scanning electron microscopy. Panel B shows a further magnified view of a region indicated in panel A.
    • Figure 6 is a series of graphs showing the results of an experiment to demonstrate that the haemolytic activity of the Ecel peptide 3 of SEQ ID NO 1 is ion-dependent. The peptide was incubated with human erythrocytes with increasing concentrations of the divalent cation chelator EDTA (A) and haemolysis assayed by measuring haemoglobin release (absorbance at 541nm, normalised to a 100% lysis control). The inhibitory effect of 50mM EDTA was reversed by the addition of 50 or 100mM magnesium (B).
    • Figure 7 is a series of graphs showing the results of experiments to investigate whether the peptide of the invention activates epithelial immunity and cFos. (A) Induction of c-Fos DNA binding activity in TR146 epithelial cells after 3 hours infection with ece1Δ/Δ null mutant, ece1Δ/Δ - ECE1 revertant and the BWP17 parent strain. (B) Production of cytokines by TR146 epithelial cells after 24 hour infection with ece1Δ/Δ null mutant, ece1Δ/Δ - ECE1 revertant, the BWP17 parent strain or uninfected cells. Data shown are the mean of 3 independent experiments. Error bars show SEM. * =p<0.05, **=p<0.01, ***=p<0.001.
    • Figure 8 shows the amino acid sequences of Ece1-III and WTSA peptides where A shows the amino acid sequence of Ece1-III (SEQ ID NO 1). Amyloidogenic regions are shown in bold type; and B shows the amino acid sequences of WTSA peptides. Each individual WTSA peptide constitutes a partial fragment of full length Ece1-III. Amyloidogenic regions are shown in bold type.
    • Figure 9 is a graph showing the results of analysis of WTSA peptide-induced damage to TR146 oral epithelial cells. Individual WTSA peptides (250 µg/ml) and WTSA 3 in combination with WTSA 6 (both peptides at 250 µg/ml) were added to confluent monolayers of TR146 oral epithelial cells and incubated for 24 hours at 37°C, 5% CO2. Following incubation, growth medium was removed and assayed for the presence of lactate dehydrogenase (LDH), a surrogate marker of cellular lysis. Dimethyl sulfoxide (DMSO) was used as a vehicle only (negative control). Full length Ece1-III (250 µg/mL) was used as the positive control.
    • Figure 10 shows WTSA peptide-mediated activation of immune responses in TR146 oral epithelial cells. WTSA peptides 1-6 were added to confluent monolayers of TR146 oral epithelial cells at a final concentration of 250 µg/mL and incubated for 24 hours at 37°C, 5% CO2. Following incubation, growth medium was removed and secreted immunostimulatory cytokines were quantified by multiplex bead assay. Concentration of detected cytokines was expressed as picograms per millilitre (pg/mL). ND= not detected.
    • Figure 11 is a series of graphs showing the results of experiments to investigate whether the peptide of the invention binds EGFR and activates c-Fos. (A) Induction of c-Fos DNA binding activity in TR146 epithelial cells after 3 hours infection with wild-type C. albicans cells, which were pre-treated for 1 hour with 4µM GW2974 (EGFR/Her2 inhibitor), 1µM PD153035 (EGFR inhibitor) or DMSO (vehicle control). (B) Sensorgram of fluid phase EGFR binding to immobilised Ece1-III of SEQ ID NO 1 demonstrating high affinity binding. Injection of fluid phase components was at point A. (C) Induction of c-Fos DNA binding activity in TR146 epithelial cells after 3 hours infection with Ece1-III, which were pre-treated for 1 hour with 4µM GW2974 (EGRF/Her2 inhibitor), 1µM PD153035 (EGFR inhibitor) or DMSO (vehicle control). Data shown are the mean of 2(A) or 3 (C) independent experiments. Error bars show SEM. * =p<0.05, **=p<0.01, ***=p<0.001.
    • Figure 12 is a set of graphs showing the effect of EGFR inhibition on epithelial cell signalling. Blocking EGFR signalling in oral epithelial cells (TR146) with Gefitnib (10µM) inhibits the activation of MAPK intracellular signalling in cells treated with Ecel-III (50µg/mL) for 2 hours as measured by decreases in levels of phosphorylated p38, JNK and ERK1/2 relative to the vehicle control (DMSO).
    • Figure 13 shows the results of an assay to detect Ecel-III-phospholipid interactions. Ecel-III was used to probe a phospholipid array (PIP Strip membrane). Binding was assessed by hybridisation with anti-Ecel-III antibody, followed by detection with anti-rabbit antibody conjugated to horse radish peroxidase.
    • Figure 14 is a series of graphs showing the results of an experiment to determine C. albicans -macrophage interactions. C. albicans strains were incubated with RAW264.7 macrophages for indicated times and the following parameters determined: (A) The number of phagocytosed fungal cells as a percentage of total number of fungal cells; (B) Hyphal length; (C) The percentage of outgrowing hyphae compared to the total number of internalised yeast mother cells.
    • Figure 15 is a graph showing the results of an experiment to show that ECE1 is required for damage of macrophages. Indicated macrophage cell types were co-incubated with C. albicans cells for 24 hours (MOI of 1 for Raws, MOI of 10 for MDMs) and macrophage damage assessed by measuring lactate dehydrogenase release into the supernatant.
    • Figure 16 shows the results of experiments showing the role of Ece1 in C. albicans mucosal pathogenesis. Infection of cortisone acetate-treated mice with wild-type (BWP17), ece1Δ null mutant or ece1Δ/Δ - ECE1 reverted strain (A) numbers of C. albicans fungi recovered per gram of kidney 3 days post-infection. Fungal invasion, tissue damage and inflammatory cell infiltration of tongue tissue 3 days post-infection by (B) BWP17, (C) ece1Δ/Δ - ECE1 and (D) ece1Δ null mutant, indicating that the ece1Δ null mutant does not invade, damage or recruit inflammatory cells. (E) tongue section 3 days post-infection with BWP17 immunostained with MPO and CD15 markers of neutrophils. ** =P<0.01.
    • Figure 17 is a graph showing the results of an experiment to show Drosophila killing by Ece1-III. The percentage of Drosophila death, 24 hours post-infection of 250µg/ml His-tagged full length Ecelp, Ecel-II(peptide 2) (Figure 2), and Ece1-III (peptide 3), into a cohort of 20 age-matched flies.
    Example 1 The role of Ecel in host cell damage
  • To investigate the role of Ecel in host cell damage, a series of C. albicans gene deletion mutants lacking ECE1, as well as revertant mutant strains were created. Specifically, C. albicans gene deletion cassettes were generated using the PCR-based method described by Dalle F et al. Cellular Microbiology 2010; 12:248-71. Primers ECE1-FG and ECE1-RG as shown below as SEQ ID NO 4 and 5 respectively, were used to amplify the HIS1 and ARG4 markers from plasmids pFA-HIS1 and pFA-ARG4, respectively. The C. albicans strain BWP17 (Wachtler B, et al. Antimicrobial Agents and Chemotherapy 2011; 55:4436-9) was sequentially transformed as described by Wachtler B, et al. PloS one 2011; 6:e17046, with the ECE1-ARG4 and ECE1-HIS1 deletion cassettes and then transformed with the Candida integrating plasmid 10 or CIp10 (Murad et al. Yeast (2000) 16, 4, p325-327), yielding the ece1Δ deletion strain. For generation of the complemented strain, the ECE1 open reading frame, plus upstream and downstream intergenic regions were amplified with primers ECE1-RecF3k (SEQ ID NO 6) and ECE1-RecR (SEQ ID NO 7) and cloned into plasmid CIp10 at MluI and SalI sites, yielding plasmid CIp10-ECE1. This plasmid was transfomed into the uridine auxotrophic ece1Δ - deletion strain, yielding the ece1Δ/Δ - ECE1 complemented strain.
  • Primers:
    • ECE1-FG
      Figure imgb0001
    • ECE1-RG
      Figure imgb0002
    • ECE1-RecF3k
      GCACGCGTCTAAAGTGGAGTAACAAC (SEQ ID NO 6)
    • ECE1-RecR
      GGTCGACCCCAGACGTTGGTTGC (SEQ ID NO 7)
  • Three strains, wild type C. albicans (wt), the ECE1 deletion mutant (ece1Δ) and the revertant mutant strain (ece1Δ/Δ - ECE1) were tested. They were applied to three independent human epithelial cell types: oral, gastrointestinal and vaginal. TR146, Caco-2 and A431 epithelial cell lines were maintained as previously described in the Dalle and Wachtler references given above. Epithelial damage assays were performed as previously described by Dalle et al. (supra.) and Wachtler et al. (2011) (supra.) with the following modifications: monolayers in 96 well plates were infected with 2 × 104 C. albicans cells; infections were performed in cell culture media without fetal bovine serum.
  • After incubation for 24 hours, the damage to the cells was assessed by measuring lactate dehydrogenase (LDH) release using a conventional assay method.
  • The results are shown in Figure 1. They show that deletion of ECE1, prevented or reduced C. albicans damage in all three cell types. As C. albicans is often found associated with the oral, gastrointestinal and urogenitary sites as a member of the natural microbiome, but can also cause infections at such sites, these data suggest that the ECE1 gene product is involved C. albicans pathogenicity.
  • Example 2 Role of Peptides
  • It has been demonstrated that Ecel can be proteolytically processed by Kex2 into eight peptide fragments (O. Bader et al. BMC Microbiology 2008, 8:115). Figure 2 illustrates the structure of Ecel and the proposed resultant peptide fragments following proteolytic processing as well as its phylogenic relationship with orthologues in C. dubliniensis and C. tropicalis (note that ECE1 orthologues are only found in these two fungal species). Each of the individual C. albicans peptides were synthesised by ProteoGenix using fmoc solid phase peptide synthesis technology.
  • The method of Example 1 was then repeated using a 1:1 mixture of peptides (1-7) of Figure 2 on human TR146 oral epithelial cells at various concentrations (250µg/ml, 125µg/ml, 62.5µg/ml, 31.25µg/ml, 15.625µg/ml and 7.8µg/ml of each individual peptide within the mixture) or peptide 3 (SEQ ID NO 1) alone in the presence of the ece1Δ mutant C. albicans. Peptide 8 could not be included, as this peptide was insoluble. After incubations for 24 hours, the damage was assessed by measuring LDH release as before. The results are shown in Figure 3A. This shows that peptide 3 (designated Ece1-III in the Figure) produced similar levels of damage to the combination at a concentration of 250µg/ml.
  • The test was then repeated using each peptide individually without C. albicans cells. The results are shown in Figure 3B and C. They show that whilst peptide 3 was capable of damaging both oral epithelial and vaginal cells, the other peptides displayed essentially no cytolytic activity. Therefore, peptide 3 of processed Ecel ("Ece1-III" of SEQ ID NO 1) is sufficient to cause lysis of human epithelial cells.
  • Example 3 Cytolytic activity of Peptides
  • In order to investigate the cytolytic activities of Ece1-III in greater detail, a haemolysis assay was carried out. Human blood was collected with the SARSTEDT blood collection system and EDTA-coated tubes, washed once with HBSS, resuspended in HBSS and stored at 4°C for up to one week for experiments. Blood was obtained from healthy human donors.
  • Each individual peptide (at a final concentration of 30µg/ml) was incubated with 1 × 107 human erythrocytes in a volume of 150µl. After incubation for 1 hour at 37°C, haemolysis was assayed by measuring haemoglobin release, as indicated by absorbance at 541nm, normalised to a 100% lysis-water control.
  • The results are shown in Figure 4. Exposure of human erythrocytes to Ece1-III, but not to the other peptides, resulted in lysis of these cells. It should be noted that, in contrast to epithelial cells, erythrocytes do not contain nuclei and cannot transcriptionally respond to stimuli. Therefore, lysis of erythrocytes by Ece1-III was due to direct cytolysis.
  • Example 4 Pore formation
  • Many peptide toxins elicit cytotoxicity via pore formation and ion influx across membranes. Whether addition of Ece1-III to epithelial cells was able to directly induce pore formation was then tested. Oral epithelial (TR-146) monolayers were incubated with 5µg/ml Ece1-III of SEQ ID NO 1 for 5 hours and pore formation assessed by scanning electron microscopy. The results are shown in Figure 5 where panel B shows a further magnified view of the region shown in panel A. Electron microscopy suggests that Ecel-III can directly induce pores (Figure 5A and B), which is in agreement with the view that Ece1-III can act as a pore-forming toxin.
  • Example 5 Ion-dependency of Haemolytic activity
  • Since the mode of action of many peptide and pore-forming toxins is ion-dependent, Ece1-III and erythrocytes were co-incubated in the presence of increasing concentrations of the divalent cation chelator, ethylenediaminetetraacetic acid (EDTA), which restricts the bioavailability of divalent cations. After 1 hour, haemolysis was measured as described in Example 3 above. The results, shown in Figure 6A, indicate that increasing concentrations of EDTA blocked the haemolytic activity of Ece1-III. This effect was reversed by supplementation with magnesium (Figure 6B). Therefore, like previously described peptide toxins, the haemolytic activity of Ece1-III appears to be ion-dependent.
  • Example 6 Activation of Immune responses
  • Pore-forming toxins are generally strong activators of inflammatory responses. Previously it has been shown that recognition of C. albicans hyphae resulted in the activation of the mitogen-activated protein kinase (MAPK) p38 signalling pathway and the c-Fos transcription factor, which in turn activates proinflammatory cytokine production (Moyes DL, et al.: Cell Host Microbe 2010, 8:225-235,). An extensive screen of >100 C. albicans mutants was carried out using protocols described in this reference and also Moyes DL, et al. PLoS ONE 2011, 6:e26580; Murciano C. et al. Infect.Immun 2011, 79:4902-4911; Moyes DL, et al. Med Microbiol Immunol 2012, 201:93-101; Moyes DL, et al. Methods MolBiol 2012, 845:345-360 and Murciano C, et al. PLoS ONE 2012, 7:e33362.
  • The results demonstrated that only a strain deleted in ECE1 was unable to activate the c-Fos pathway (Figure 7A) or proinflammatory cytokines (Figure 7B) from oral epithelial cells whilst still producing hyphae. Therefore, activation of epithelial immune responses via c-Fos against C. albicans is the result of the action and/or recognition of Ecel.
  • Example 7 Functional requirements of Ece1-III
  • Having identified Ece1-III as the region of Ecel responsible for cellular lysis and the activation of immune responses, the applicants next investigated the precise functional requirements of the Ece1-III amino acid sequence (Figure 8A). Accordingly, individual peptide fragments corresponding to internal regions of Ece1-III were constructed (WTSA peptides 1-6 (SEQ ID NOS 8-11, 3 and 12 respectively: Figure 8B). The sequence of each WTSA peptide was chosen to facilitate a detailed examination of each of the internal amyloidogenic regions of Ece1-III, either alone ( WTSA peptides 1, 2, 4 and 5), or in combination (WTSA 3). An additional fragment of Ece1-III lacking both amyloidogenic regions was also created (WTSA 6: Figure 8B). The WTSA peptides were analysed for their ability to cause epithelial cell lysis. However, none of the individual WTSA peptide fragments were observed to induce cell lysis (Figure 9). Notably, when WTSA 3 and 6 were applied to epithelial cells in combination, (which together constitute the entire Ecel-III peptide sequence) the ability to induce cell lysis was not restored. The WTSA peptides were then analysed for their ability to stimulate secretion of pro-inflammatory cytokines from oral epithelial cells. None of the WTSA peptides stimulated significant levels of cytokine secretion (Figure 10). In contrast, a potent induction of all cytokines was observed following exposure to full length Ece1-III. Taken together, these data indicate that only intact, full length Ece1-III is capable of inducing cellular lysis and stimulation of the inflammatory response in epithelial cells.
  • The p38/c-Fos pathway is activated via the interaction of C. albicans hyphae with the surface receptor EGFR (epidermal growth factor receptor), as blocking EGFR activation also blocks c-Fos activation (Figure 11A). Biacore binding assays show that Ece1-III (of SEQ ID NO 1), but not other parts of the Ecel protein, directly interact with EGFR with high affinity (Figure 11B) and that blocking EGFR abolishes the ability of Ece1-III to activate c-Fos (Figure 11C). Furthermore, blocking EGFR also reduced/abolished activation of all three MAPK signalling pathways (p38, JNK and ERK1/2) (Fig. 12) This demonstrates that activation of mucosal immune responses via the p38/c-Fos pathway is the result of Ecel-III/EGFR interactions and that EGFR plays a primary role in the activation of cell signalling by Ece1-III.
  • Example 8 Interaction of Ece1-III with Phospholipids
  • In order to insert into a target membrane, a pore-forming peptide toxin must interact with components of the membrane. Because Ece1-III consists of a predicted alpha helix with a dibasic (lysine, arginine) C-terminal head, the applicants predicted that these cationic, positively charged residues may interact with negatively charged phospholipids present in target membranes. Ece1-III was used to probe a phospholipid array (PIP Strip membrane). Binding was assessed by hybridisation with anti-Ece1-III antibody, followed by detection with anti-rabbit antibody conjugated to horse radish peroxidase. The results are shown in Figure 13. These show that Ece1-III interacts with Phosphatidylinositol-3-phosphate, Phosphatidylinositol-4-phosphate, Phosphatidylinositol-5-phosphate, Phosphatidylinositol-3,4-phosphate, Phosphatidylinositol-3,5 -phosphate, Phosphatidylinositol-4,5-phosphate, Phosphatidylinositol-3,4,5-phosphate, Phosphatidic acid and Phosphatidylserine, with binding to Phosphatidylinositol-3-phosphate being particularly robust (Figure 13). Ece1-III did not interact with Lysophosphatidic acid, Lysophosphatidylcholine, Phosphatidylinositol, Phosphatidylethanolamine, Phosphatidylcholine or Sphingosine-1-phosphate. Therefore, insertion of Ece1-III into target membranes (pore formation) may be mediated by interactions with phospholipids.
  • Example 9 Ecel facilitates immune cell killing
  • Given the importance of Ece1-III in killing epithelial cells, the applicants examined whether it also plays a role in immune escape of C. albicans from macrophages. In vitro, C. albicans yeast cells are phagocytosed by macrophages, but rapidly germinate and escape the macrophage via hypha formation. To examine whether Ecel is involved in this process, wild type, ece1Δ, and ece1Δ/Δ - ECE1 cells were coincubated with macrophages and the following parameters measured: phagocytosis rates, length of hyphae, escape from macrophages, and damage of macrophages. Interestingly, ece1Δ cells were phagocytosed by macrophages at the same rate, formed hyphae within macrophages of the same length, and even escaped from macrophages at the same rates as the wild type, but were unable to damage these immune cells (Figure 14 and 15). These data demonstrate that C. albicans hyphae can non-lytically escape from macrophages, and indicate a specific role for Ecel in host cell damage.
  • Example 10 Effect of Ecel on mucosal pathosenciity in vivo
  • The applicants also tested whether Ecel was required for mucosal pathogenciity in vivo. To test this, they utilised a murine model of oropharyngeal candidiasis. In this model, mice were first immunosuppressed with cortisone acetate prior to administration of wild-type (parental) C. albicans (BWP17), a ece1Δ null mutant and ece1Δ/Δ - ECE1 revertant strain into the oral cavity. After four days, fungal burdens, fungal invasion, tissue damage and immune cell recruitment was determined in tongue tissues. Significantly lower fungal burdens were found in mice infected with the ece1Δ null mutant as compared with the wild-type (BWP17) or the ece1Δ/Δ - ECE1 revertant strains, and in many cases the ece1Δ null mutant could not be recovered from mice (Figure 16 A). Histological analysis of mouse tongues in both the wild-type (BWP17) and ece1Δ/Δ - ECE1 revertant strain showed multiple foci of infection that were associated with inflammatory immune cells (neutrophils) and extensive local tissue damage (Figure 16 B-D). In contrast, mice infected with the ece1Δ null mutant showed no evidence of any foci of invasion, tissue damage or inflammation and no evidence of C. albicans was detectable (Figure 16 E). Therefore, Ecel is essential for successful mucosal infection, damage induction and immune activation in this model.
  • Example 11 Effect of Ecel and Ece1-III on Drosophila melanogaster
  • The applicants also determined the effect of administering full length His-tagged Ecelp and Ece1-III in a Drosophila melanogaster (fruit fly) model. All flies injected with full length His-tagged Ecelp and Ece1-III displayed signs of paralysis, with only 17.5% and 10% recovering after 24 hours, as compared with the negative control Ecel-II (peptide 2) which showed no signs of paralysis (Figure 17). This indicates that Ece1-III may have a direct or indirect neurotoxic effect.
  • Conclusion
  • These data provide evidence that Ecel (and its active component Ece1-III) has a dual functional role, by acting both as a pore-forming toxin that damages host cells and as an activator of immune responses. Damage of host cells and uncontrolled immune activation are the hallmarks of several diseases caused by C. albicans, which can often lead to death (45- 76% mortality) during systemic infections. Therefore, neutralisation of Ece1-III will prevent not only host damage during C. albicans infections but also deleterious inflammatory responses. This indicates that Ece1-III represents a new therapeutic target to treat C. albicans infections.
  • SEQUENCE LISTING
    • <110> King's College London
      Leibniz Institute fur Naturstoff-Forschung und Infektionshbioligie e.v.
    • <120> Peptides and Binding Partners Therefor
    • <130> P3010/WO
    • <150> GB1306588.3
      <151> 2013-04-11
    • <160> 15
    • <170> BiSSAP 1.2
    • <210> 1
      <211> 32
      <212> PRT
      <213> Candida albicans
    • <220>
      <223> Peptide
    • <400> 1
      Figure imgb0003
    • <210> 2
      <211> 9
      <212> PRT
      <213> Candida albicans
    • <220>
      <223> Peptide
    • <400> 2
      Figure imgb0004
    • <210> 3
      <211> 10
      <212> PRT
      <213> Candida albicans
    • <220>
      <223> Peptide
    • <400> 3
      Figure imgb0005
    • <210> 4
      <211> 122
      <212> DNA
      <213> Artificial Sequence
    • <220>
      <221> source
      <222> 1..122
      <223> /mol_type="unassigned DNA" /note="Primer" /organism="Artificial Sequence"
    • <400> 4
      Figure imgb0006
    • <210> 5
      <211> 127
      <212> DNA
      <213> Artificial Sequence
    • <220>
      <221> source
      <222> 1..127
      <223> /mol_type="unassigned DNA" /note="Primer" /organism="Artificial Sequence"
    • <400> 5
      Figure imgb0007
    • <210> 6
      <211> 26
      <212> PRT
      <213> Artificial Sequence
    • <220>
      <223> Primer
    • <400> 6
      Figure imgb0008
    • <210> 7
      <211> 23
      <212> DNA
      <213> Artificial Sequence
    • <220>
      <221> source
      <222> 1..23
      <223> /mol_type="unassigned DNA" /note="Primer" /organism="Artificial Sequence"
    • <400> 7
      ggtcgacccc agacgttggt tgc   23
    • <210> 8
      <211> 10
      <212> PRT
      <213> Candida albicans
    • <220>
      <223> Peptide
    • <400> 8
      Figure imgb0009
    • <210> 9
      <211> 14
      <212> PRT
      <213> Candida albicans
    • <220>
      <223> Peptide
    • <400> 9
      Figure imgb0010
    • <210> 10
      <211> 24
      <212> PRT
      <213> Candida albicans
    • <220>
      <223> Peptide
    • <400> 10
      Figure imgb0011
    • <210> 11
      <211> 14
      <212> PRT
      <213> Candida albicans
    • <220>
      <223> Peptide
    • <400> 11
      Figure imgb0012
    • <210> 12
      <211> 8
      <212> PRT
      <213> Candida albicans
    • <220>
      <223> Peptide
    • <400> 12
      Figure imgb0013
    • <210> 13
      <211> 271
      <212> PRT
      <213> Candida albicans
    • <220>
      <223> Ecel
    • <400> 13
      Figure imgb0014
    • <210> 14
      <211> 268
      <212> PRT
      <213> Candida dubliniensis
    • <220>
      <223> Ecel
    • <400> 14
      Figure imgb0015
      Figure imgb0016
    • <210> 15
      <211> 282
      <212> PRT
      <213> Candida tropicalis
    • <220>
      <223> Ecel
    • <400> 15
      Figure imgb0017
      Figure imgb0018

Claims (14)

  1. A peptide which is of SEQ ID NO 1
    SIIGIIMGILGNIPQVIQIIMSIVKAFKGNKR (SEQ ID NO.1) or a variant thereof wherein the variant is peptide encoded by an orthologue of a gene sequence which encodes the peptide of SEQ ID NO 1 derived from an Ecel protein of a Candida species, or a peptide which is at least 87% identical to SEQ ID No 1; or a fragment of either of these of from 20 to 31 amino acids in length; or a labelled form thereof, wherein the peptide has a cell-activating, damage-inducing, immunomodulatory or immunogenic ability.
  2. A peptide according to claim 1 which comprises at least one of SEQ ID NO.2 or SEQ ID NO 3: IIGIIMGIL (SEQ ID NO. 2) QVIQIIMSIV (SEQ ID NO. 3).
  3. A peptide according to claim 1 or claim 2 which is of SEQ ID NO 1.
  4. A peptide according to claim 1 which is a peptide encoded by an orthologue of a gene sequence from C. dubliniensis of SEQ ID NO 16
    SIIGILTAILNNVPQIINVITTIIKSIITGNKR (SEQ ID NO. 16)
    or from C. tropicalis of SEQ ID NO 17
    ISFAGIVSSIINQLPSIIQIIGNIIKAGLVKR (SEQ ID NO. 17).
  5. A peptide according to any one of the preceding claims which is a fragment which lacks the dibasic (KR) C-terminal head.
  6. A specific binding partner for a peptide according to any one of claims 1 to 4, or a labelled form thereof, wherein the specific binding partner is an antibody or a binding fragment thereof, for use in the treatment or prevention of infection by Candida species.
  7. A pharmaceutical composition comprising a peptide according to any one of claims 1 to 4 or a specific binding partner according to claim 6 and a pharmaceutically acceptable carrier.
  8. A peptide according to any one of claims 1 to 4 for use in therapy, such as for the treatment or prevention of infection by Candida albicans.
  9. A nucleic acid that encodes a peptide according to any one of claims 1 to 4.
  10. A recombinant cell that has been transformed with a nucleic acid according to claim 9.
  11. A method for producing a peptide to any one of claims 1 to 4, which method comprises culturing a recombinant cell according to claim 10 and recovering said peptide therefrom.
  12. A method for diagnosis of an infection by C. albicans, said method comprising contacting a sample which has been obtained from an individual with a peptide according to any one of claims 1 to 4, and detecting whether the sample contains moieties that interact with said peptide at a sufficient level to indicate a pathogenic invasive infection.
  13. A kit for diagnosing an infection by C. albicans, said kit comprising a peptide according to any one of claims 1 to 4, and means for detecting complexes formed between said peptide and a specific binding partner.
  14. An in vitro method for screening for compounds having anti-fungal activity, said method comprising determining whether a test compound will interact with a peptide according to any one of claims 1 to 4.
EP14722312.7A 2013-04-11 2014-04-10 Peptides and binding partners therefor Active EP2984103B1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1306588.3A GB201306588D0 (en) 2013-04-11 2013-04-11 Peptides and Binding Partners Therefor
PCT/GB2014/051118 WO2014167335A1 (en) 2013-04-11 2014-04-10 Peptides and binding partners therefor

Publications (2)

Publication Number Publication Date
EP2984103A1 EP2984103A1 (en) 2016-02-17
EP2984103B1 true EP2984103B1 (en) 2019-06-12

Family

ID=48537092

Family Applications (1)

Application Number Title Priority Date Filing Date
EP14722312.7A Active EP2984103B1 (en) 2013-04-11 2014-04-10 Peptides and binding partners therefor

Country Status (5)

Country Link
US (2) US9969796B2 (en)
EP (1) EP2984103B1 (en)
CN (1) CN105377883A (en)
GB (1) GB201306588D0 (en)
WO (1) WO2014167335A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201306588D0 (en) 2013-04-11 2013-05-29 King S College London Peptides and Binding Partners Therefor
WO2020130838A2 (en) * 2018-12-21 2020-06-25 Qvq Holding B.V. Antibodies for preventing or treating candidiasis

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6747137B1 (en) * 1998-02-13 2004-06-08 Genome Therapeutics Corporation Nucleic acid sequences relating to Candida albicans for diagnostics and therapeutics
EP1235918B1 (en) 1999-11-29 2007-03-21 Trustees of Dartmouth College Methods for altering the expression of hyphal-specific genes
WO2006036817A2 (en) * 2004-09-24 2006-04-06 Microbia, Inc. Fungal variants and uses thereof
MX2012000018A (en) 2009-07-03 2012-03-29 Los Angeles Biomed Res Inst Hyr1 as a target for active and passive immunization against candida.
GB201306588D0 (en) 2013-04-11 2013-05-29 King S College London Peptides and Binding Partners Therefor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
US20160046699A1 (en) 2016-02-18
US20180215812A1 (en) 2018-08-02
WO2014167335A1 (en) 2014-10-16
CN105377883A (en) 2016-03-02
EP2984103A1 (en) 2016-02-17
GB201306588D0 (en) 2013-05-29
US9969796B2 (en) 2018-05-15

Similar Documents

Publication Publication Date Title
US7241613B1 (en) Identification of Candida cell surface proteins and their uses
EP2998740A1 (en) Method for predicting clinical effect of immunotherapy
WO2005061532A1 (en) Pathogenic infection detection compositions and methods
US10806776B2 (en) Method of treating fungal infection
US20180215812A1 (en) Peptides and binding partners therefor
KR20100028558A (en) Pseudomonas aeruginosa outer membrane protein pa4710
JP2000502070A (en) Treatment and diagnosis of infections by Helicobacter pylori
CN101454446A (en) Pseudomonas aeruginosa outer membrane protein PA0427
CN110476065B (en) Diagnostic method
US20170198021A1 (en) Methods of treating diabetes and compositions capable of same
WO2007049770A1 (en) Outer coat protein pa5158 of pseudomonas aeruginosa
US11655274B2 (en) Glycosylated YghJ polypeptides from enterotoxigenic Escherichia coli (ETEC)
AU684009B2 (en) Peptides for diagnostics
US11492391B1 (en) Intracellular nanobody targeting T4SS effector inhibits ehrlichia infection
US20240192209A1 (en) Diagnosis of bartonella using recombinant proteins
EP2396655B1 (en) Device for serological investigation of yersinia infections and/or subsequent illnesses and use of proteins MyfA and PsaA of Y enterocolitica and Y. pseudotuberculosis as recombinant antigens
NL2023527B1 (en) Peptides for use in the treatment of cholera
WO2015154783A1 (en) Glycosylated polypeptides originating from enterotoxigenic escherichia coli (etec)
CN115724940A (en) Targeted STAT3 protein Tyr705 phosphorylation peptide and application thereof
Emrick Characterization of the fungicidal activity and biochemical impact of occidiofungin, a novel antifungal compound derived from Burkholderia contaminans
WO2012126172A1 (en) Peptides inhibiting adhesion of aspergillus fumigatus to cornea
CN110958886A (en) Plasmodium falciparum and plasmodium vivax vaccines
Li et al. E03-08-0618, Revised January 8, 2004 Clustering and redistribution of late endocytic compartments in response to Helicobacter pylori vacuolating toxin

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20151023

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RIN1 Information on inventor provided before grant (corrected)

Inventor name: HUBE, BERNHARD

Inventor name: NAGLIK, JULIAN

Inventor name: MOYES, DAVID

Inventor name: WILSON, DUNCAN

Inventor name: RICHARDSON, JONATHAN

Inventor name: TANG, SHIRLEY

Inventor name: HOFS, SARAH

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20170131

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20180918

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTC Intention to grant announced (deleted)
INTG Intention to grant announced

Effective date: 20190307

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1142433

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190615

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602014048169

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: LATSCHA SCHOELLHORN PARTNER AG, CH

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190912

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190913

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190912

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1142433

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191014

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191012

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602014048169

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

26N No opposition filed

Effective date: 20200313

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200224

PG2D Information on lapse in contracting state deleted

Ref country code: IS

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200410

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200410

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190612

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230519

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602014048169

Country of ref document: DE

Representative=s name: MAIWALD GMBH, DE

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20240422

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20240411

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20240418

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20240501

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20240417

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20240419

Year of fee payment: 11